heme oxygenase-1 as a therapeutic target in inflammatory disorders of the gastrointestinal tract

8
Vijith Vijayan, Eveline Baumgart-Vogt, Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, 35392 Gies- sen, Germany Sebastian Mueller, Salem Medical Center and Center of Al- cohol Research, University of Heidelberg, 69121 Heidelberg, Germany Stephan Immenschuh, Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany Author contributions: Vijayan V and Immenschuh S con- tributed to manuscript conception, preparation and writing; Baumgart-Vogt E and Mueller S carried out manuscript review. Supported by Grant SFB547 A8 from the Deutsche Forschun- gsgemeinschaft (to Immenschuh S) Correspondence to: Stephan Immenschuh, MD, Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg- Str. 1, 30625 Hannover, Germany. [email protected] Telephone: +49-511-5326704 Fax: +49-511-5322079 Received: February 22, 2010 Revised: April 1, 2010 Accepted: April 8, 2010 Published online: July 7, 2010 Abstract Heme oxygenase (HO)-1 is the inducible isoform of the first and rate-limiting enzyme of heme degradation. HO-1 not only protects against oxidative stress and apoptosis, but has received a great deal of attention in recent years because of its potent anti-inflammatory functions. Studies with HO-1 knockout animal models have led to major advances in the understanding of how HO-1 might regulate inflammatory immune responses, although little is known on the underlying mechanisms. Due to its beneficial effects the targeted induction of this enzyme is considered to have major therapeutic po- tential for the treatment of inflammatory disorders. This review discusses current knowledge on the mechanisms that mediate anti-inflammatory protection by HO-1. More specifically, the article deals with the role of HO-1 in the pathophysiology of viral hepatitis, inflammatory bowel disease, and pancreatitis. The effects of specific HO-1 modulation as a potential therapeutic strategy in experimental cell culture and animal models of these gastrointestinal disorders are summarized. In conclu- sion, targeted regulation of HO-1 holds major promise for future clinical interventions in inflammatory diseases of the gastrointestinal tract. © 2010 Baishideng. All rights reserved. Key words: Antioxidant; Heme oxygenase; Hepatitis; Immunity; Inflammation; Inflammatory bowel disease; Oxidative stress; Pancreatitis Peer reviewer: Dr. Rene Schmidt, PhD, Department of An- esthesiology, Freiburg University Medical Center, Hugstetter Strasse 55, 79106 Freiburg, Germany Vijayan V, Mueller S, Baumgart-Vogt E, Immenschuh S. Heme oxygenase-1 as a therapeutic target in inflammatory disor- ders of the gastrointestinal tract. World J Gastroenterol 2010; 16(25): 3112-3119 Available from: URL: http://www.wjgnet. com/1007-9327/full/v16/i25/3112.htm DOI: http://dx.doi. org/10.3748/wjg.v16.i25.3112 INTRODUCTION Heme oxygenase (HO), which was initially described more than 40 years ago, enzymatically degrades heme and produces equimolar amounts of carbon monoxide (CO), biliverdin, and iron [1] (Figure 1). In a coupled reaction, biliverdin is converted into bilirubin (BR) via biliverdin reductase [2] . Two distinct isozymes of HO, HO-1 and HO-2, have been identified and represent the products of two different genes with distinct tissue- and cell-specific expression patterns [3-5] . The constitutive isoform HO-2 is preferentially expressed in brain and testis [6] , and is es- sentially not regulated by metabolic or receptor-mediated 3112 World J Gastroenterol 2010 July 7; 16(25): 3112-3119 ISSN 1007-9327 (print) © 2010 Baishideng. All rights reserved. Online Submissions: http://www.wjgnet.com/1007-9327office [email protected] doi:10.3748/wjg.v16.i25.3112 July 7, 2010|Volume 16|Issue 25| WJG|www.wjgnet.com Heme oxygenase-1 as a therapeutic target in inflammatory disorders of the gastrointestinal tract Vijith Vijayan, Sebastian Mueller, Eveline Baumgart-Vogt, Stephan Immenschuh TOPIC HIGHLIGHT Kostas Pantopoulos, Associate Professor, Series Editor

Upload: independent

Post on 29-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Vijith Vijayan, Eveline Baumgart-Vogt, Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, 35392 Gies-sen, GermanySebastian Mueller, Salem Medical Center and Center of Al-cohol Research, University of Heidelberg, 69121 Heidelberg, GermanyStephan Immenschuh, Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, GermanyAuthor contributions: Vijayan V and Immenschuh S con-tributed to manuscript conception, preparation and writing; Baumgart-Vogt E and Mueller S carried out manuscript review.Supported by Grant SFB547 A8 from the Deutsche Forschun-gsgemeinschaft (to Immenschuh S)Correspondence to: Stephan Immenschuh, MD, Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany. [email protected]: +49-511-5326704 Fax: +49-511-5322079Received: February 22, 2010 Revised: April 1, 2010Accepted: April 8, 2010Published online: July 7, 2010

AbstractHeme oxygenase (HO)-1 is the inducible isoform of the first and rate-limiting enzyme of heme degradation. HO-1 not only protects against oxidative stress and apoptosis, but has received a great deal of attention in recent years because of its potent anti-inflammatory functions. Studies with HO-1 knockout animal models have led to major advances in the understanding of how HO-1 might regulate inflammatory immune responses, although little is known on the underlying mechanisms. Due to its beneficial effects the targeted induction of this enzyme is considered to have major therapeutic po-tential for the treatment of inflammatory disorders. This review discusses current knowledge on the mechanisms that mediate anti-inflammatory protection by HO-1. More specifically, the article deals with the role of HO-1 in the pathophysiology of viral hepatitis, inflammatory

bowel disease, and pancreatitis. The effects of specific HO-1 modulation as a potential therapeutic strategy in experimental cell culture and animal models of these gastrointestinal disorders are summarized. In conclu-sion, targeted regulation of HO-1 holds major promise for future clinical interventions in inflammatory diseases of the gastrointestinal tract.

© 2010 Baishideng. All rights reserved.

Key words: Antioxidant; Heme oxygenase; Hepatitis; Immunity; Inflammation; Inflammatory bowel disease; Oxidative stress; Pancreatitis

Peer reviewer: Dr. Rene Schmidt, PhD, Department of An-esthesiology, Freiburg University Medical Center, Hugstetter Strasse 55, 79106 Freiburg, Germany

Vijayan V, Mueller S, Baumgart-Vogt E, Immenschuh S. Heme oxygenase-1 as a therapeutic target in inflammatory disor-ders of the gastrointestinal tract. World J Gastroenterol 2010; 16(25): 3112-3119 Available from: URL: http://www.wjgnet.com/1007-9327/full/v16/i25/3112.htm DOI: http://dx.doi.org/10.3748/wjg.v16.i25.3112

INTRODUCTIONHeme oxygenase (HO), which was initially described more than 40 years ago, enzymatically degrades heme and produces equimolar amounts of carbon monoxide (CO), biliverdin, and iron[1] (Figure 1). In a coupled reaction, biliverdin is converted into bilirubin (BR) via biliverdin reductase[2]. Two distinct isozymes of HO, HO-1 and HO-2, have been identified and represent the products of two different genes with distinct tissue- and cell-specific expression patterns[3-5]. The constitutive isoform HO-2 is preferentially expressed in brain and testis[6], and is es-sentially not regulated by metabolic or receptor-mediated

3112

World J Gastroenterol 2010 July 7; 16(25): 3112-3119 ISSN 1007-9327 (print)

© 2010 Baishideng. All rights reserved.

Online Submissions: http://www.wjgnet.com/[email protected]:10.3748/wjg.v16.i25.3112

July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Heme oxygenase-1 as a therapeutic target in inflammatory disorders of the gastrointestinal tract

Vijith Vijayan, Sebastian Mueller, Eveline Baumgart-Vogt, Stephan Immenschuh

TOPIC HIGHLIGHT

Kostas Pantopoulos, Associate Professor, Series Editor

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

stimuli[4,7]. In contrast, the inducible HO isozyme, HO-1, which exhibits low basal expression levels in most cells and tissues, is markedly upregulated not only by its sub-strate heme, but also by other oxidative stress stimuli, such as UV-light and lipopolysaccharide (LPS) or directly by reactive oxygen species, such as hydrogen peroxide. In addition, heavy metals, sulfhydryl-reactive reagents, and hypoxia are potent inducers of HO-1[8-10]. Thus, although HO-1 does not directly catalyze an antioxidant reaction, its induction is generally considered an adaptive cytoprotec-tive response against the toxicity of oxidative stress[11-13]. In the current review, we focus our attention on recent findings that show the emerging anti-inflammatory and immunomodulatory role of HO-1 and its products. In particular, we highlight recent advances in the understand-ing how HO-1 might modulate the inflammatory immune response and the potential role of HO-1 for therapeutic applications in inflammatory conditions of various organs in the gastrointestinal tract.

HO-1 GENE DEFICIENCY CAUSES

PROINFLAMMATORY PHENOTYPICAL

ALTERATIONSA potential link between HO-1 and inflammatory disease has been shown by Willis et al[14] in an animal model of carragenin-induced pleurisy, in which specific upregula-tion of HO enzyme activity attenuated a complement-de-pendent inflammation. Mice that are deficient for HO-1 not only develop chronic inflammation, but are also highly vulnerable to experimental sepsis induced by the classical proinflammatory mediator endotoxin[15]. In ad-dition, innate and adaptive immune reactions are severely affected in these knockout mice[16-18]. In contrast, HO-2 knockout mice appear to have an intact immune regula-tion, but exhibit defects in their central and autonomous nervous system[19]. The phenotype of the only known human case of genetic HO-1 deficiency is very similar to that observed in HO-1 knockout mice. This HO-1 defi-cient patient, a Japanese boy who died at the age of six years, was initially diagnosed with anemia and a chronic inflammatory disorder[20,21].

ANTI-INFLAMMATORY EFFECTS OF HO-1 IN MONONUCLEAR PHAGOCYTES AND ENDOTHELIAL CELLS Although HO-1 is expressed in all tissues and cells, the im-munomodulatory functions of HO-1 appear to be primar-ily dependent on HO-1 functions in mononuclear phago-cytes and endothelial cells. In the following, we describe pertinent findings that illustrate the anti-inflammatory role of HO-1 in these two cell types.

Mononuclear phagocytesMononuclear phagocytes, such as monocytes and mac-

rophages, are cells with a common bone marrow lineage and have versatile functions in the innate and adaptive immune system[22,23]. As an example, macrophages ingest and kill invading microorganisms as a first line of defence and are activated by various immunological stimuli, such as microbial products or cytokines. In response to these stimuli, macrophages are able to initiate and enhance the inflammatory immune response[23]. It has been known for many years that tissue macrophages, such as Kupffer cells and spleen macrophages, are cell types in which HO-1 is highly expressed under normal conditions[24,25]. Several studies have been performed with cultured mononuclear cells from rodents. Here, HO-1 is upregulated in response to LPS[25-27], which attenuates the expression of various proinflammatory genes, such as cyclooxygenase-2, tu-mor necrosis factor-α, interleukin (IL)-1, and IL-6[18,28-31]. The cell-specific antiinflammatory function of HO-1 in mononuclear phagocytes was recently confirmed in a con-ditional HO-1 knockout mouse model. The cell-specific lack of HO-1 in mononuclear cells caused a major defect of interferon-β expression. In addition, a pathological immune response in an experimental infection with Sen-dai virus and in autoimmune encephalomyelitis in these animals was observed[32]. Interestingly, others have shown that HO-1 is also important for the appropriate function of human and mouse dendritic cells. It has been shown that pharmacological induction of HO-1 attenuates matu-ration and cell-specific functions of dendritic cells[33,34]. Moreover, HO-1 expression in mononuclear cells has been demonstrated to be essential for the functionality of regulatory T cells[17].

Endothelial cellsThe endothelium plays a central role in the regulation of inflammatory reactions, because it serves as a barrier between the peripheral blood stream and inflamed tis-sues. More specifically, endothelial monolayers regulate the recruitment and transmigration of immunologically active blood cells, such as polymorphonuclear leukocytes (neutrophils) or T lymphocytes, to the site of an inflam-mation[28,35,36]. Hayashi and colleagues[37] have reported that HO-1 regulates cell-cell interactions between poly-morphonuclear leukocytes and endothelial cells. These authors showed in an in vivo rat model that the increased

3113 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Heme

Heme oxygenase

CO iron NADP

Biliverdin

Biliverdinreductase

Bilirubin

NADPH

Figure 1 The heme oxygenase enzyme reaction. Heme is enzymatically de-graded to yield carbon monoxide (CO), iron, and biliverdin, which is converted into bilirubin in a coupled reaction.

enzyme activity of HO in the endothelium of microves-sels downregulated the adhesion of leukocytes during experimental oxidative stress[37]. Accordingly, others have demonstrated that the activity of endothelial HO-1 spe-cifically modulates leukocyte recruitment into organs with an experimental inflammation[38]. Independently, in a streptozocin-induced rat model of experimental diabetes, overexpression of HO-1 has been shown to attenuate oxi-dative stress-dependent endothelial cell damage[39]. Similar findings in the endothelium have recently been reported for knockout mice, in which genetic deficiency of HO-1 caused major pathological alterations of the endothelial monolayer[40]. Specifically, endothelial cells from HO-1 de-ficient mice were shown to be more susceptible to apop-tosis and denudation from the extracellular matrix. More-over, independent groups have shown that antiinflamma-tory endothelial protection of HO-1 might be mediated via its ability to downregulate the tumor necrosis factor-α-induced expression of various adhesion molecules[41-43]. It is also remarkable that histopathological studies in the autopsy of a human patient with genetic HO-1 deficiency revealed major endothelial cell damage[21].

HOW DOES HO-1 MEDIATE ITS ANTIINFLAMMATORY FUNCTIONSThe mechanisms of how HO-1 may counteract inflam-matory reactions are not understood in detail. An ac-cumulating body of evidence, however, indicates that the HO substrate heme is a compound with major pro-inflammatory properties and that the HO products BR and CO have potent antiinflammatory functions.

Heme as a proinflammatory compoundThe tetrapyrrole heme has contradictory biological prop-erties. On the one hand, heme is important for oxygen and mitochondrial electron transport as the prosthetic group of various hemoproteins such as hemoglobin, myo-globin, and cytochromes[44,45]. On the other hand, heme can be toxic as it can cause oxidative stress in its “free” non-protein bound form. The prooxidant properties of heme have been shown in various animal and cell culture models[46,47]. Due to the critical role of intracellular heme levels, enzymatic synthesis and degradation of this com-pound is tightly controlled[48-50]. More recently, heme has also been recognized to exhibit potent proinflammatory properties[18,51]. As an example, Jeney et al[52] have dem-onstrated that heme-dependent oxidation of low-density lipoproteins is involved in inflammation-mediated tissue damage. In this report, the proinflammatory effects of heme have also been shown to be correlated with specific clinical conditions, such as atherosclerosis and hemolytic anemia[52]. Independently, others have demonstrated that intravenous administration of heme caused experimental inflammation in vivo with a major influx of leukocytes[38]. Heme-dependent toxicity has also been associated with its proinflammatory effects in an animal model of experi-

mental cerebral malaria, in which heme-dependent detri-mental effects were markedly more pronounced in HO-1 knockout mice[53]. Recent findings might shed light on the mechanisms that could be involved in the proinflammato-ry effects of heme. Figueiredo and colleagues have dem-onstrated that initiation of heme-dependent inflammation was mediated via specific interaction of heme with the central pattern recognition receptor toll-like receptor-4, in a cell culture model of mononuclear cells and in mice[54]. The role of HO-1 in maintaining intracellular heme ho-meostasis has been reviewed elsewhere[18,47,48,51].

BilirubinThe role of bilirubin (BR) as a beneficial compound with potent antioxidant and antiinflammatory effects has only been appreciated in recent years[55,56]. Protection against an experimental inflammation has been shown for HO-derived BR in an animal model[37]. Moreover, it has been observed in a murine asthma model that BR specifically reduces leukocyte transmigration to the site of an ex-perimental inflammation via interaction with the adhe-sion molecule vascular cell adhesion molecule-1[57]. More recently, it has been reported in a mouse model of sepsis that a single bolus of BR markedly blocked the toxicity of endotoxin[58]. Epidemiological studies have indicated that moderately increased concentrations of serum BR (e.g. in Gilbert’s disease) are associated with a lower risk of developing cardiovascular disease[59,60] and colorectal car-cinoma[61]. In conclusion, the generation of BR by HO-1 might, in part, explain the antiinflammatory effects of this enzyme.

COAlthough CO is generally considered a toxic gas, vari-ous physiological functions of CO as a major signaling molecule have been recognized in recent years[3,62,63]. In particular, HO-1-derived CO has been shown to be in-volved in the regulation of apoptosis, neurotransmission, coagulation, vasodilation, and inflammation. In a pio-neering report on the potential protective effects of this gas, administration of exogenous CO has been shown to block the LPS-induced production of proinflammatory cytokines via modulation of p38 MAP kinase[29]. Similar to the signaling gas NO, CO upregulates the production of cGMP via activation of soluble guanyl cyclase. This mechanism has also been implicated in other functions of CO, such as vasodilation and blockage of smooth muscle cell proliferation. A remarkable development with major potential for future therapeutic applications has been the introduction of CO-releasing molecules (CORMs). CORMs are compounds that can be admin-istered intravenously and are intended to deliver CO to its target site without the toxicity of gaseous CO[64]. Further details on CO and CORMs are given in specific reviews[63,65,66].

HO-derived ironIron, as the third product of HO, is an essential com-

3114 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

pound for redox-dependent enzyme reactions and bioen-ergetics. Iron, however, might cause the formation of tox-ic reactive oxygen species, if not appropriately contained by specific intracellular protective mechanisms. A key role in the protection against the toxicity of HO-1-derived iron is played by the intracellular iron storage protein ferri-tin[67,68]. This protection has been demonstrated in various cell culture models, in which the synthesis of HO-1 and ferritin was coordinately upregulated and prevented iron-mediated cell toxicity[12,47,69]. Remarkably, genetic deficiency of HO-1 in mice[70] and humans[20,21] is associated with a major pathological iron overload in the liver and kidney.

In summary, the HO products BR and CO have anti-inflammatory potential, which could be relevant for thera-peutic interventions.

TARGETING HO-1 IN INFLAMMATORY DISORDERS OF THE GASTROINTESTINAL TRACT HO-1 and chronic viral hepatitisChronic viral hepatitis mainly consists of viral hepatitis B and C, and is a major cause of liver cirrhosis and end-stage liver disease worldwide. These disorders are charac-terized by chronic self-perpetuating inflammation of the liver for at least six months. For example, chronic hepatitis C virus (HCV) infection affects 4 million US citizens, a third of whom will progress to liver cirrhosis and primary hepatocellular carcinoma if left untreated[71-73]. Treatment with interferon and ribavirin is effective in only 50% of patients, because many patients either have contraindica-tions to these therapies or have failed to respond to this treatment, indicating a need for alternative or supplemen-tary therapeutic strategies. In the following, we will focus on links between HO-1 and chronic viral hepatitis and their potential therapeutic use. Research on hepatitis B virus (HBV) and HCV infections has been prevented by a lack of appropriate animal and cell culture models to study these infections[73].

As regards HO-1 and HBV infection, Protzer and col-

leagues have reported that HO-1 has antiviral activity in a transgenic mouse model of chronic HBV infection. In this animal model, specific induction of HO-1 by cobalt-protoporphyrin-IX significantly reduced the levels of the viral HBV core protein. Moreover, these authors have shown that the antiviral effects of HO-1 are mediated via reduction of HBV core protein stability in cell cultures of stably transfected hepatoma cells[74] (Table 1).

Several reports suggest that HO-1 might serve as a specific therapeutic target for the treatment of chronic HCV infection, although the results are somewhat con-tradictory. On the one hand, Schmidt and colleagues have presented evidence that HCV specifically downregulates gene expression of HO-1, but not that of other antioxi-dant genes, such as manganese superoxide dismutase and catalase[75]. In cell cultures of hepatoma cells, inhibition of HO-1 by HCV appeared to be specifically regulated via the HCV-core protein[75,76]. On the other hand, it has been demonstrated that HCV leads to an increased expression of HO-1 in hepatoma cell lines, possibly via interactions with the HO-1 repressor, Bach1[77]. Clearly, further studies are necessary to reconcile these conflicting results.

In contrast, findings from independent reports have indicated that targeted overexpression of HO-1 had anti-viral effects on HCV replication (Table 1). The group of Bonkovsky has demonstrated that upregulation of HO-1 either by a pharmacological approach or, more recently, by a microRNA-based strategy repressed HCV replication in human Huh-7 hepatoma cells[78,79]. Accordingly, Zhu et al[80] have shown that targeted overexpression of HO-1 led to a significant inhibition of HCV replication without affect-ing other parameters of cell viability in human hepatoma cells, which stably replicate subgenomic selectable HCV RNAs[80]. These studies have recently been extended by others, who have shown that the HO-1 product biliverdin interfered with HCV replication via direct modulation of the antiviral interferon-α response in two human hepa-toma replicon cell lines[81] (Table 1).

Due to the limited success rate of current therapies for chronic viral hepatitis, it is conceivable that targeted modulation of HO-1 might be an innovative comple-

3115 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Table 1 HO-1 and inflammatory disorders of the gastrointestinal tract

Disorder Mechanism of protection by HO-1 Experimental model Ref.

Viral hepatitis(HBV, HCV)

HBV: repression of HBV replication In vivo: transgenic mice; in vitro: human HepG2 hepatoma cells [74]

HCV: repression of HCV replication In vitro: human Huh-7 hepatoma cells [78-80]

In vitro: human Huh-5-15 hepatoma cells [81]

Inflammatory bowel disease

HO-1-derived biliverdin inhibits inflammatory activity In vivo: colitis in dextran sodium sulfate-treated mice [89]

Inhibition of IRF8 activation In vivo: colitis in IL-10-/- mice [90]

HO-1 mediates protection of 5-aminosalicylic acid In vivo: colitis in trinitrobenzene sulfonic acid-treated rats [91]

Inhibition of NF-kB activation In vivo: colitis in trinitrobenzene sulfonic acid-treated mice [92]

Inhibition of interleukin-17 In vivo: colitis in dextran sodium sulfate-treated mice [93]

Pancreatitis Homing of hemin-treated macrophages in pancreas before onset of inflammation

In vivo: acute pancreatitis in choline-deficient diet-fed mice [101]

Decreased expression of proinflammatory cytokines In vivo: acute pancreatitis after allograft transplantation in rat [102]

Inhibition of cell proliferation via repression of ERK activity In vitro: platelet-derived growth-factor-treated rat pancreatic stellate cells

[105]

HO: Heme oxygenase; HBV: Hepatitis B virus; HCV: Hepatitis C virus.

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

mentary strategy for the treatment of these infectious diseases.

HO-1 and inflammatory bowel diseaseInflammatory bowel disease (IBD) is a group of chronic inflammatory disorders, which is primarily represented by ulcerative colitis and Crohn’s disease. The etiology of IBD is not well understood and a complex interplay of genetic, immunological and environmental factors appears to play a role in the initiation and perpetuation of IBD[82]. The onset of IBD is characterized by an autoimmune inflam-matory reaction that causes excessive production of pro-inflammatory cytokines and reactive oxygen species, which in turn damage the intestinal mucosa[83]. A number of ex-perimental animal models of IBD have been established, in which IBD develops either spontaneously, such as in various knockout mouse models, or after treatment with specific chemical compounds, such as trinitrobenzene sul-fonic acid[84]. These models are widely used to investigate potential therapeutic strategies for the treatment of IBD. HO-1 regulation has been studied in a variety of these ani-mal models and in biopsies from human IBD patients.

In a trinitrobenzene sulfonic acid-induced model of experimental colitis, it has been demonstrated that HO-1 is prominently upregulated in various cell types of the inflamed colon[85]. These findings correspond with ob-servations from an independent study, in which increased HO-1 expression has been observed in mucosa biopsies of a murine colitis model. Importantly, in this study, HO-1 expression has also been shown to be increased in inflamed mucosa of human IBD patients[86]. Similar find-ings have been reported by Takagi et al[87] for a Japanese population. Indirect evidence that HO-1 might play a role in the pathogenesis of IBD has been demonstrated in knockout mice for the Nrf2 gene, which is the major transcriptional regulator of HO-1. Nrf2-deficient mice were more susceptible to develop an experimental colitis in response to dextran sodium sulphate when compared with their wild-type counterparts[88].

As regards the potential therapeutic role of HO-1 in IBD, important insights have been presented in inde-pendent reports on HO products (Table 1). In a dextran sodium sulphate-induced colitis model, HO-1-dependent protection against inflammation has been attributed to the beneficial effects of biliverdin[89]. By contrast, others have indicated that CO might provide anti-inflammatory protection in mice with genetic IL-10 deficiency, which develop a chronic colitis-like disease. In these mice it is also shown that the immunosuppressive effects of CO were recapitulated by pharmacological induction of HO-1[90]. Interestingly, upregulation of HO-1 might also play a role in the antiinflammatory protective effects of some established current standard therapies of IBD. As an example, Horváth et al[91] have shown that 5-amino sali-cylic acid (5-ASA), which is one of the pharmacological standard therapies of IBD, might, at least in part, mediate its antiinflammatory effects via upregulation of HO-1 in a trinitrobenzene sulfonic acid-induced rat colitis model. In

addition, others have demonstrated that the fungal metab-olite gliotoxin and the HO-1 substrate heme mediate an-tiinflammatory effects in independent experimental colitis models via specific induction of HO-1, respectively[92,93] (Table 1).

Finally, it is important to point out that targeted HO-1 induction does not provide anti-inflammatory protec-tion in an experimental animal model of dextran sulphate sodium-induced colitis, when HO-1 was induced after the onset of IBD[86]. Therefore, it seems questionable whether HO-1 induction is useful for treatment of established IBD, but rather might be useful as a preventive measure.

HO-1 and pancreatitisPancreatitis is an inflammatory disorder that is clinically categorized into acute and chronic pancreatitis[94]. The clinical stages of pancreatitis range from a transient self-limiting inflammatory reaction to a fulminant disease with necrotic lesions. Severe acute pancreatitis, which might have multiple local and systemic complications, is associ-ated with high mortality[87]. In general, acute pancreatitis is caused by alcohol abuse and gallstones. More recently, however, genetics and obesity have been identified as in-dependent risk factors. The pathogenesis of pancreatitis is not well understood, but it is known that proinflammatory pancreas-independent factors, such as exposure to endo-toxin, can trigger the onset of the disease. Oxidative stress has repeatedly been implicated in the pathogenesis of pancreatitis[95,96]. Although the exact role of oxidative stress during the course of the disease is not well understood, antioxidant enzymes and vitamins have been shown to improve the clinical consequences of pancreatitis[97]. Other therapeutic strategies for the treatment of pancreatitis in-clude inactivation of pancreatic enzymes and blockage of platelet factor activating receptor[94,97,98].

HO-1 is upregulated in animal models of experimental pancreatitis in a cell-specific manner[99,100]. In particular, it has been demonstrated that HO-1 gene expression in the inflamed pancreas is primarily upregulated in peripheral macrophages, which migrate into areas of inflammation[101] (Table 1). In the latter report, the authors have also asked whether HO-1 induction might protect against pancreati-tis. The major finding of this study, in which experimental pancreatitis was induced with a choline-deficient diet, indi-cated that administration of heme decreased pancreatitis-associated lethality in mice. Moreover, administration of heme increased the pancreatic tissue secretion of chemo-kines, which was responsible for the infiltration of HO-1 expressing peritoneal macrophages into the pancreas. It is important to note, however, that heme-dependent HO-1 induction after the onset of pancreatitis failed to reduce the severity of the disease. This finding is similar to what has been observed in IBD, as mentioned above. Thus, al-though HO-1 induction might not be useful for the treat-ment of established pancreatitis, it might help to prevent the onset of pancreatitis in a clinical setting, in which pan-creatitis is likely to develop. In a model of pancreas trans-plantation, in which pancreatitis develops as a consequence

3116 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

of ischemia-reperfusion injury, pharmacological induction of HO-1 has been shown to be protective against pan-creatitis[102] (Table 1). Similar observations have also been reported in an independent study, in which pretreatment with cobalt-protoporphyrin-IX prevented the microcir-culatory dysfunction after pancreas ischemia-reperfusion injury in rat[103]. More recently, the CO donor CORM-2 has been shown to protect against acute pancreatitis in rats[104].

Finally, specific induction of HO-1 might also be ap-plicable for treatment of chronic pancreatitis. In a recent report, Schwer et al[105] have shown that HO-1 induction by curcumin inhibited pancreatic stellate cell proliferation, which plays a major role in the pathogenesis of pancreatic fibrosis in chronic pancreatitis. This protection was abol-ished by blockage of HO activity, either by an enzyme in-hibitor or by knockdown of HO-1 with a specific siRNA (Table 1).

CONCLUSIONTargeted overexpression of HO-1 has major potential for the treatment of inflammatory disorders of the gastroin-testinal tract. Current knowledge on the applications of HO-1 as a therapeutic target, however, still seems preco-cious and critical questions remain to be answered before clinical interventions might be available.

REFERENCES1 Tenhunen R, Marver HS, Schmid R. The enzymatic conver-

sion of heme to bilirubin by microsomal heme oxygenase. Proc Natl Acad Sci USA 1968; 61: 748-755

2 Kapitulnik J, Maines MD. Pleiotropic functions of biliver-din reductase: cellular signaling and generation of cytopro-tective and cytotoxic bilirubin. Trends Pharmacol Sci 2009; 30: 129-137

3 Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol 1997; 37: 517-554

4 Ryter SW, Alam J, Choi AM. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev 2006; 86: 583-650

5 Abraham NG, Kappas A. Pharmacological and clinical as-pects of heme oxygenase. Pharmacol Rev 2008; 60: 79-127

6 Trakshel GM, Kutty RK, Maines MD. Purification and char-acterization of the major constitutive form of testicular heme oxygenase. The noninducible isoform. J Biol Chem 1986; 261: 11131-11137

7 Cruse I, Maines MD. Evidence suggesting that the two forms of heme oxygenase are products of different genes. J Biol Chem 1988; 263: 3348-3353

8 Choi AM, Alam J. Heme oxygenase-1: function, regula-tion, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Cell Mol Biol 1996; 15: 9-19

9 Immenschuh S, Ramadori G. Gene regulation of heme oxy-genase-1 as a therapeutic target. Biochem Pharmacol 2000; 60: 1121-1128

10 Alam J, Igarashi K, Immenschuh S, Shibahara S, Tyrrell RM. Regulation of heme oxygenase-1 gene transcription: recent advances and highlights from the International Conference (Uppsala, 2003) on Heme Oxygenase. Antioxid Redox Signal 2004; 6: 924-933

11 Keyse SM, Applegate LA, Tromvoukis Y, Tyrrell RM. Oxi-dant stress leads to transcriptional activation of the human

heme oxygenase gene in cultured skin fibroblasts. Mol Cell Biol 1990; 10: 4967-4969

12 Vile GF, Basu-Modak S, Waltner C, Tyrrell RM. Heme oxy-genase 1 mediates an adaptive response to oxidative stress in human skin fibroblasts. Proc Natl Acad Sci USA 1994; 91: 2607-2610

13 Otterbein LE, Choi AM. Heme oxygenase: colors of defense against cellular stress. Am J Physiol Lung Cell Mol Physiol 2000; 279: L1029-L1037

14 Willis D, Moore AR, Frederick R, Willoughby DA. Heme oxygenase: a novel target for the modulation of the inflam-matory response. Nat Med 1996; 2: 87-90

15 Poss KD, Tonegawa S. Reduced stress defense in heme oxygenase 1-deficient cells. Proc Natl Acad Sci USA 1997; 94: 10925-10930

16 Kapturczak MH, Wasserfall C, Brusko T, Campbell-Thomp-son M, Ellis TM, Atkinson MA, Agarwal A. Heme oxygen-ase-1 modulates early inflammatory responses: evidence from the heme oxygenase-1-deficient mouse. Am J Pathol 2004; 165: 1045-1053

17 George JF, Braun A, Brusko TM, Joseph R, Bolisetty S, Was-serfall CH, Atkinson MA, Agarwal A, Kapturczak MH. Sup-pression by CD4+CD25+ regulatory T cells is dependent on expression of heme oxygenase-1 in antigen-presenting cells. Am J Pathol 2008; 173: 154-160

18 Soares MP, Marguti I, Cunha A, Larsen R. Immunoregula-tory effects of HO-1: how does it work? Curr Opin Pharmacol 2009; 9: 482-489

19 Burnett AL, Johns DG, Kriegsfeld LJ, Klein SL, Calvin DC, Demas GE, Schramm LP, Tonegawa S, Nelson RJ, Snyder SH, Poss KD. Ejaculatory abnormalities in mice with targeted disruption of the gene for heme oxygenase-2. Nat Med 1998; 4: 84-87

20 Yachie A, Niida Y, Wada T, Igarashi N, Kaneda H, Toma T, Ohta K, Kasahara Y, Koizumi S. Oxidative stress causes en-hanced endothelial cell injury in human heme oxygenase-1 deficiency. J Clin Invest 1999; 103: 129-135

21 Kawashima A, Oda Y, Yachie A, Koizumi S, Nakanishi I. Heme oxygenase-1 deficiency: the first autopsy case. Hum Pathol 2002; 33: 125-130

22 Gordon S. The macrophage. Bioessays 1995; 17: 977-98623 Zhang X, Mosser DM. Macrophage activation by endog-

enous danger signals. J Pathol 2008; 214: 161-17824 Bissell DM, Hammaker L, Schmid R. Liver sinusoidal cells.

Identification of a subpopulation for erythrocyte catabolism. J Cell Biol 1972; 54: 107-119

25 Immenschuh S, Tan M, Ramadori G. Nitric oxide mediates the lipopolysaccharide dependent upregulation of the heme oxygenase-1 gene expression in cultured rat Kupffer cells. J Hepatol 1999; 30: 61-69

26 Immenschuh S, Stritzke J, Iwahara S, Ramadori G. Up-regulation of heme-binding protein 23 (HBP23) gene expres-sion by lipopolysaccharide is mediated via a nitric oxide-dependent signaling pathway in rat Kupffer cells. Hepatol-ogy 1999; 30: 118-127

27 Wijayanti N, Huber S, Samoylenko A, Kietzmann T, Im-menschuh S. Role of NF-kappaB and p38 MAP kinase sig-naling pathways in the lipopolysaccharide-dependent acti-vation of heme oxygenase-1 gene expression. Antioxid Redox Signal 2004; 6: 802-810

28 Immenschuh S, Schröder H. Heme oxygenase-1 and cardio-vascular disease. Histol Histopathol 2006; 21: 679-685

29 Otterbein LE, Bach FH, Alam J, Soares M, Tao Lu H, Wysk M, Davis RJ, Flavell RA, Choi AM. Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nat Med 2000; 6: 422-428

30 Lin HY, Juan SH, Shen SC, Hsu FL, Chen YC. Inhibition of lipopolysaccharide-induced nitric oxide production by fla-vonoids in RAW264.7 macrophages involves heme oxygen-ase-1. Biochem Pharmacol 2003; 66: 1821-1832

31 Itoh K, Mochizuki M, Ishii Y, Ishii T, Shibata T, Kawamoto

3117 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

Y, Kelly V, Sekizawa K, Uchida K, Yamamoto M. Transcrip-tion factor Nrf2 regulates inflammation by mediating the effect of 15-deoxy-Delta(12,14)-prostaglandin j(2). Mol Cell Biol 2004; 24: 36-45

32 Tzima S, Victoratos P, Kranidioti K, Alexiou M, Kollias G. Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-beta production. J Exp Med 2009; 206: 1167-1179

33 Chauveau C, Rémy S, Royer PJ, Hill M, Tanguy-Royer S, Hubert FX, Tesson L, Brion R, Beriou G, Gregoire M, Josien R, Cuturi MC, Anegon I. Heme oxygenase-1 expression inhib-its dendritic cell maturation and proinflammatory function but conserves IL-10 expression. Blood 2005; 106: 1694-1702

34 Rémy S, Blancou P, Tesson L, Tardif V, Brion R, Royer PJ, Motterlini R, Foresti R, Painchaut M, Pogu S, Gregoire M, Bach JM, Anegon I, Chauveau C. Carbon monoxide inhib-its TLR-induced dendritic cell immunogenicity. J Immunol 2009; 182: 1877-1884

35 Muller WA. Leukocyte-endothelial-cell interactions in leukocyte transmigration and the inflammatory response. Trends Immunol 2003; 24: 327-334

36 Cook-Mills JM, Deem TL. Active participation of endothe-lial cells in inflammation. J Leukoc Biol 2005; 77: 487-495

37 Hayashi S, Takamiya R, Yamaguchi T, Matsumoto K, Tojo SJ, Tamatani T, Kitajima M, Makino N, Ishimura Y, Suemat-su M. Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress: role of bili-rubin generated by the enzyme. Circ Res 1999; 85: 663-671

38 Wagener FA, Eggert A, Boerman OC, Oyen WJ, Verhofstad A, Abraham NG, Adema G, van Kooyk Y, de Witte T, Figdor CG. Heme is a potent inducer of inflammation in mice and is counteracted by heme oxygenase. Blood 2001; 98: 1802-1811

39 Abraham NG, Rezzani R, Rodella L, Kruger A, Taller D, Li Volti G, Goodman AI, Kappas A. Overexpression of human heme oxygenase-1 attenuates endothelial cell sloughing in experimental diabetes. Am J Physiol Heart Circ Physiol 2004; 287: H2468-H2477

40 True AL, Olive M, Boehm M, San H, Westrick RJ, Ragha-vachari N, Xu X, Lynn EG, Sack MN, Munson PJ, Gladwin MT, Nabel EG. Heme oxygenase-1 deficiency accelerates formation of arterial thrombosis through oxidative damage to the endothelium, which is rescued by inhaled carbon mon-oxide. Circ Res 2007; 101: 893-901

41 Vachharajani TJ, Work J, Issekutz AC, Granger DN. Heme oxygenase modulates selectin expression in different re-gional vascular beds. Am J Physiol Heart Circ Physiol 2000; 278: H1613-H1617

42 Liu X, Spolarics Z. Methemoglobin is a potent activator of endothelial cells by stimulating IL-6 and IL-8 produc-tion and E-selectin membrane expression. Am J Physiol Cell Physiol 2003; 285: C1036-C1046

43 Soares MP, Seldon MP, Gregoire IP, Vassilevskaia T, Ber-berat PO, Yu J, Tsui TY, Bach FH. Heme oxygenase-1 modu-lates the expression of adhesion molecules associated with endothelial cell activation. J Immunol 2004; 172: 3553-3563

44 Padmanaban G, Venkateswar V, Rangarajan PN. Haem as a multifunctional regulator. Trends Biochem Sci 1989; 14: 492-496

45 Wijayanti N, Katz N, Immenschuh S. Biology of heme in health and disease. Curr Med Chem 2004; 11: 981-986

46 Vincent SH. Oxidative effects of heme and porphyrins on proteins and lipids. Semin Hematol 1989; 26: 105-113

47 Ryter SW, Tyrrell RM. The heme synthesis and degradation pathways: role in oxidant sensitivity. Heme oxygenase has both pro- and antioxidant properties. Free Radic Biol Med 2000; 28: 289-309

48 Ponka P. Cell biology of heme. Am J Med Sci 1999; 318: 241-256

49 Sassa S. Novel effects of heme and heme-related compounds in biological systems. Curr Med Chem 1996; 3: 273-290

50 Sassa S. Why heme needs to be degraded to iron, biliverdin

IXalpha, and carbon monoxide? Antioxid Redox Signal 2004; 6: 819-824

51 Wagener FA, Volk HD, Willis D, Abraham NG, Soares MP, Adema GJ, Figdor CG. Different faces of the heme-heme oxygenase system in inflammation. Pharmacol Rev 2003; 55: 551-571

52 Jeney V, Balla J, Yachie A, Varga Z, Vercellotti GM, Eaton JW, Balla G. Pro-oxidant and cytotoxic effects of circulating heme. Blood 2002; 100: 879-887

53 Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, Epiphanio S, Chora A, Rodrigues CD, Gregoire IP, Cunha-Rodrigues M, Portugal S, Soares MP, Mota MM. Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimen-tal cerebral malaria. Nat Med 2007; 13: 703-710

54 Figueiredo RT, Fernandez PL, Mourao-Sa DS, Porto BN, Dutra FF, Alves LS, Oliveira MF, Oliveira PL, Graça-Souza AV, Bozza MT. Characterization of heme as activator of Toll-like receptor 4. J Biol Chem 2007; 282: 20221-20229

55 Stocker R. Antioxidant activities of bile pigments. Antioxid Redox Signal 2004; 6: 841-849

56 Kapitulnik J. Bilirubin: an endogenous product of heme degradation with both cytotoxic and cytoprotective proper-ties. Mol Pharmacol 2004; 66: 773-779

57 Keshavan P, Deem TL, Schwemberger SJ, Babcock GF, Cook-Mills JM, Zucker SD. Unconjugated bilirubin inhibits VCAM-1-mediated transendothelial leukocyte migration. J Immunol 2005; 174: 3709-3718

58 Kadl A, Pontiller J, Exner M, Leitinger N. Single bolus injec-tion of bilirubin improves the clinical outcome in a mouse model of endotoxemia. Shock 2007; 28: 582-588

59 Mayer M. Association of serum bilirubin concentration with risk of coronary artery disease. Clin Chem 2000; 46: 1723-1727

60 Schwertner HA, Vítek L. Gilbert syndrome, UGT1A1*28 allele, and cardiovascular disease risk: possible protective effects and therapeutic applications of bilirubin. Atheroscle-rosis 2008; 198: 1-11

61 Zucker SD, Horn PS, Sherman KE. Serum bilirubin levels in the U.S. population: gender effect and inverse correlation with colorectal cancer. Hepatology 2004; 40: 827-835

62 Boczkowski J, Poderoso JJ, Motterlini R. CO-metal interac-tion: Vital signaling from a lethal gas. Trends Biochem Sci 2006; 31: 614-621

63 Kim HP, Ryter SW, Choi AM. CO as a cellular signaling molecule. Annu Rev Pharmacol Toxicol 2006; 46: 411-449

64 Motterlini R, Clark JE, Foresti R, Sarathchandra P, Mann BE, Green CJ. Carbon monoxide-releasing molecules: char-acterization of biochemical and vascular activities. Circ Res 2002; 90: E17-E24

65 Motterlini R, Mann BE, Foresti R. Therapeutic applications of carbon monoxide-releasing molecules. Expert Opin Inves-tig Drugs 2005; 14: 1305-1318

66 Foresti R, Bani-Hani MG, Motterlini R. Use of carbon mon-oxide as a therapeutic agent: promises and challenges. In-tensive Care Med 2008; 34: 649-658

67 Ponka P. Tissue-specific regulation of iron metabolism and heme synthesis: distinct control mechanisms in erythroid cells. Blood 1997; 89: 1-25

68 Papanikolaou G, Pantopoulos K. Iron metabolism and tox-icity. Toxicol Appl Pharmacol 2005; 202: 199-211

69 Balla G, Jacob HS, Balla J, Rosenberg M, Nath K, Apple F, Ea-ton JW, Vercellotti GM. Ferritin: a cytoprotective antioxidant strategem of endothelium. J Biol Chem 1992; 267: 18148-18153

70 Poss KD, Tonegawa S. Heme oxygenase 1 is required for mammalian iron reutilization. Proc Natl Acad Sci USA 1997; 94: 10919-10924

71 Sallie R, Di Bisceglie AM. Viral hepatitis and hepatocellular carcinoma. Gastroenterol Clin North Am 1994; 23: 567-579

72 Goodman ZD, Ishak KG. Histopathology of hepatitis C vi-rus infection. Semin Liver Dis 1995; 15: 70-81

3118 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders

73 Hoofnagle JH. Course and outcome of hepatitis C. Hepatol-ogy 2002; 36: S21-S29

74 Protzer U, Seyfried S, Quasdorff M, Sass G, Svorcova M, Webb D, Bohne F, Hösel M, Schirmacher P, Tiegs G. Anti-viral activity and hepatoprotection by heme oxygenase-1 in hepatitis B virus infection. Gastroenterology 2007; 133: 1156-1165

75 Abdalla MY, Ahmad IM, Spitz DR, Schmidt WN, Britigan BE. Hepatitis C virus-core and non structural proteins lead to different effects on cellular antioxidant defenses. J Med Virol 2005; 76: 489-497

76 Abdalla MY, Britigan BE, Wen F, Icardi M, McCormick ML, LaBrecque DR, Voigt M, Brown KE, Schmidt WN. Down-regulation of heme oxygenase-1 by hepatitis C virus infec-tion in vivo and by the in vitro expression of hepatitis C core protein. J Infect Dis 2004; 190: 1109-1118

77 Ghaziani T, Shan Y, Lambrecht RW, Donohue SE, Pi-etschmann T, Bartenschlager R, Bonkovsky HL. HCV pro-teins increase expression of heme oxygenase-1 (HO-1) and decrease expression of Bach1 in human hepatoma cells. J Hepatol 2006; 45: 5-12

78 Shan Y, Zheng J, Lambrecht RW, Bonkovsky HL. Recipro-cal effects of micro-RNA-122 on expression of heme oxy-genase-1 and hepatitis C virus genes in human hepatocytes. Gastroenterology 2007; 133: 1166-1174

79 Hou W, Tian Q, Zheng J, Bonkovsky HL. MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expres-sion in human hepatoma cells expressing hepatitis C viral proteins. Hepatology 2010; 51: 1494-1504

80 Zhu Z, Wilson AT, Mathahs MM, Wen F, Brown KE, Luxon BA, Schmidt WN. Heme oxygenase-1 suppresses hepatitis C virus replication and increases resistance of hepatocytes to oxidant injury. Hepatology 2008; 48: 1430-1439

81 Lehmann E, El-Tantawy WH, Ocker M, Bartenschlager R, Lohmann V, Hashemolhosseini S, Tiegs G, Sass G. The heme oxygenase 1 product biliverdin interferes with hepa-titis C virus replication by increasing antiviral interferon response. Hepatology 2010; 51: 398-404

82 Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med 2009; 361: 2066-2078

83 Deban L, Correale C, Vetrano S, Malesci A, Danese S. Mul-tiple pathogenic roles of microvasculature in inflammatory bowel disease: a Jack of all trades. Am J Pathol 2008; 172: 1457-1466

84 Wirtz S, Neurath MF. Animal models of intestinal inflam-mation: new insights into the molecular pathogenesis and immunotherapy of inflammatory bowel disease. Int J Colorectal Dis 2000; 15: 144-160

85 Wang WP, Guo X, Koo MW, Wong BC, Lam SK, Ye YN, Cho CH. Protective role of heme oxygenase-1 on trinitro-benzene sulfonic acid-induced colitis in rats. Am J Physiol Gastrointest Liver Physiol 2001; 281: G586-G594

86 Paul G, Bataille F, Obermeier F, Bock J, Klebl F, Strauch U, Lochbaum D, Rümmele P, Farkas S, Schölmerich J, Fleck M, Rogler G, Herfarth H. Analysis of intestinal haem-oxygen-ase-1 (HO-1) in clinical and experimental colitis. Clin Exp Immunol 2005; 140: 547-555

87 Takagi T, Naito Y, Mizushima K, Nukigi Y, Okada H, Su-zuki T, Hirata I, Omatsu T, Okayama T, Handa O, Kokura S, Ichikawa H, Yoshikawa T. Increased intestinal expression of heme oxygenase-1 and its localization in patients with ulcer-ative colitis. J Gastroenterol Hepatol 2008; 23 Suppl 2: S229-S233

88 Khor TO, Huang MT, Kwon KH, Chan JY, Reddy BS, Kong AN. Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium-induced colitis. Cancer Res 2006; 66: 11580-11584

89 Berberat PO, A-Rahim YI, Yamashita K, Warny MM, Csiz-

madia E, Robson SC, Bach FH. Heme oxygenase-1-gener-ated biliverdin ameliorates experimental murine colitis. Inflamm Bowel Dis 2005; 11: 350-359

90 Hegazi RA, Rao KN, Mayle A, Sepulveda AR, Otterbein LE, Plevy SE. Carbon monoxide ameliorates chronic murine colitis through a heme oxygenase 1-dependent pathway. J Exp Med 2005; 202: 1703-1713

91 Horváth K, Varga C, Berkó A, Pósa A, László F, Whittle BJ. The involvement of heme oxygenase-1 activity in the thera-peutic actions of 5-aminosalicylic acid in rat colitis. Eur J Pharmacol 2008; 581: 315-323

92 Jun CD, Kim Y, Choi EY, Kim M, Park B, Youn B, Yu K, Choi KS, Yoon KH, Choi SC, Lee MS, Park KI, Choi M, Chung Y, Oh J. Gliotoxin reduces the severity of trinitro-benzene sulfonic acid-induced colitis in mice: evidence of the connection between heme oxygenase-1 and the nuclear factor-kappaB pathway in vitro and in vivo. Inflamm Bowel Dis 2006; 12: 619-629

93 Zhong W, Xia Z, Hinrichs D, Rosenbaum JT, Wegmann KW, Meyrowitz J, Zhang Z. Hemin exerts multiple protective mechanisms and attenuates dextran sulfate sodium-induced colitis. J Pediatr Gastroenterol Nutr 2010; 50: 132-139

94 Forsmark CE, Baillie J. AGA Institute technical review on acute pancreatitis. Gastroenterology 2007; 132: 2022-2044

95 Schoenberg MH, Büchler M, Beger HG. The role of oxygen radicals in experimental acute pancreatitis. Free Radic Biol Med 1992; 12: 515-522

96 Sweiry JH, Mann GE. Role of oxidative stress in the patho-genesis of acute pancreatitis. Scand J Gastroenterol Suppl 1996; 219: 10-15

97 Hofbauer B, Saluja AK, Lerch MM, Bhagat L, Bhatia M, Lee HS, Frossard JL, Adler G, Steer ML. Intra-acinar cell activa-tion of trypsinogen during caerulein-induced pancreatitis in rats. Am J Physiol 1998; 275: G352-G362

98 Chen C, Xia SH, Chen H, Li XH. Therapy for acute pancre-atitis with platelet-activating factor receptor antagonists. World J Gastroenterol 2008; 14: 4735-4738

99 Sato H, Siow RC, Bartlett S, Taketani S, Ishii T, Bannai S, Mann GE. Expression of stress proteins heme oxygenase-1 and -2 in acute pancreatitis and pancreatic islet betaTC3 and acinar AR42J cells. FEBS Lett 1997; 405: 219-223

100 Fu K, Sarras MP Jr, De Lisle RC, Andrews GK. Expression of oxidative stress-responsive genes and cytokine genes during caerulein-induced acute pancreatitis. Am J Physiol 1997; 273: G696-G705

101 Nakamichi I, Habtezion A, Zhong B, Contag CH, Butcher EC, Omary MB. Hemin-activated macrophages home to the pancreas and protect from acute pancreatitis via heme oxy-genase-1 induction. J Clin Invest 2005; 115: 3007-3014

102 Becker T, Zu Vilsendorf AM, Terbish T, Klempnauer J, Jörns A. Induction of heme oxygenase-1 improves the sur-vival of pancreas grafts by prevention of pancreatitis after transplantation. Transplantation 2007; 84: 1644-1655

103 von Dobschuetz E, Schmidt R, Scholtes M, Thomusch O, Schwer CI, Geiger KK, Hopt UT, Pannen BH. Protective role of heme oxygenase-1 in pancreatic microcirculatory dys-function after ischemia/reperfusion in rats. Pancreas 2008; 36: 377-384

104 Chen P, Sun B, Chen H, Wang G, Pan S, Kong R, Bai X, Wang S. Effects of carbon monoxide releasing molecule-liberated CO on severe acute pancreatitis in rats. Cytokine 2010; 49: 15-23

105 Schwer CI, Guerrero AM, Humar M, Roesslein M, Goebel U, Stoll P, Geiger KK, Pannen BH, Hoetzel A, Schmidt R. Heme oxygenase-1 inhibits the proliferation of pancreatic stellate cells by repression of the extracellular signal-regulated ki-nase1/2 pathway. J Pharmacol Exp Ther 2008; 327: 863-871

S- Editor Tian L L- Editor Stewart GJ E- Editor Ma WH

3119 July 7, 2010|Volume 16|Issue 25|WJG|www.wjgnet.com

Vijayan V et al . Heme oxygenase-1 as a therapeutic target in inflammatory disorders